Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cells ; 11(17)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36078135

RESUMEN

Peripheral adiponectin acts on the hypothalamus to inhibit energy expenditure and increase food intake through its receptors AdipoR1 and adipoR2. The hypothalamic expression of adiponectin is poorly documented. We hypothesize that whether hypothalamic adiponectin is confirmed, its expression and secretion could be regulated as peripheral adiponectin. Thus, in the present work, we aim to determine whether adiponectin is expressed in the hypothalamus and in two neuronal cell lines and investigate the potential mechanisms regulating its neuronal expression. Using immunohistochemistry, we show that adiponectin is expressed in the mediobasal hypothalamic neurons of mice. Adiponectin expression is also evidenced in two neuronal cell lines mHypo POMC (an adult mouse hypothalamic cell line) and SH-SY5Y (human neuroblastoma). The neuronal expression of adiponectin is increased in response to rosiglitazone treatment (a PPARγ agonist) and FGF21 and is decreased in insulin-resistant neurons. Furthermore, we show that adiponectin expressed by mHypo POMC neurons is secreted in a culture medium. Adiponectin also diminished the resistin-induced IL6 expression in SIMA9 cells, a microglia cell line. In conclusion, we evidenced the hypothalamic expression of adiponectin and its regulation at the neuronal level.


Asunto(s)
Adiponectina , Neuronas , Adiponectina/metabolismo , Adulto , Animales , Humanos , Ratones , Neuroblastoma/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptores de Adiponectina/metabolismo
2.
Mol Cell Endocrinol ; 533: 111341, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34082045

RESUMEN

Resistin has been firstly discovered in mice and was identified as an adipose tissue-secreted hormone or adipokine linking obesity and insulin resistance. In humans, resistin has been characterized as a hormone expressed and secreted by Immune cells especially by macrophages, and was linked to many inflammatory responses including inflammation of adipose tissue due to macrophages' infiltration. Human and mouse resistin display sequence and structural similarities and also dissimilarities that could explain their different expression pattern. In mice, strong pieces of evidence clearly associated high resistin plasma levels to obesity and insulin resistance suggesting that resistin could play an important role in the onset and progression of obesity and insulin resistance via resistin-induced inflammation. In humans, the link between resistin and obesity/insulin resistance is still a matter of debate and needs more epidemiological studies. Also, resistin has been linked to other chronic diseases such as cardiovascular diseases and cancers where resistin has been proposed in many studies as a biological marker.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Enfermedades Metabólicas/metabolismo , Neoplasias/metabolismo , Resistina/metabolismo , Animales , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Enfermedades Metabólicas/genética , Ratones , Neoplasias/genética , Resistina/genética , Especificidad de la Especie
3.
Sci Rep ; 11(1): 5427, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33686181

RESUMEN

Saturated fatty acids such as palmitic acid promote inflammation and insulin resistance in peripheral tissues, contrasting with the protective action of polyunsaturated fatty acids such docosahexaenoic acid. Palmitic acid effects have been in part attributed to its potential action through Toll-like receptor 4. Beside, resistin, an adipokine, also promotes inflammation and insulin resistance via TLR4. In the brain, palmitic acid and resistin trigger neuroinflammation and insulin resistance, but their link at the neuronal level is unknown. Using human SH-SY5Yneuroblastoma cell line we show that palmitic acid treatment impaired insulin-dependent Akt and Erk phosphorylation whereas DHA preserved insulin action. Palmitic acid up-regulated TLR4 as well as pro-inflammatory cytokines IL6 and TNFα contrasting with DHA effect. Similarly to palmitic acid, resistin treatment induced the up-regulation of IL6 and TNFα as well as NFκB activation. Importantly, palmitic acid potentiated the resistin-dependent NFkB activation whereas DHA abolished it. The recruitment of TLR4 to membrane lipid rafts was increased by palmitic acid treatment; this is concomitant with the augmentation of resistin-induced TLR4/MYD88/TIRAP complex formation mandatory for TLR4 signaling. In conclusion, palmitic acid increased TLR4 expression promoting resistin signaling through TLR4 up-regulation and its recruitment to membrane lipid rafts.


Asunto(s)
Resistencia a la Insulina , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Ácido Palmítico/farmacología , Resistina/metabolismo , Línea Celular Tumoral , Humanos , Inflamación/metabolismo
4.
Metabolism ; 123: 154846, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34371064

RESUMEN

Oestrogens regulate body weight through their action on hypothalamus to modulate food intake and energy expenditure. Hypothalamic de novo ceramide synthesis plays a central role on obesity induced by oestrogen deficiency. Depletion in oestrogens is also known to be associated with glucose intolerance, which favours type 2 diabetes (T2D). However, the implication of hypothalamic ceramide in the regulation of glucose homeostasis by oestrogen is unknown. Here, we studied glucose homeostasis and insulin secretion in ovariectomized (OVX) female rats. OVX induces body weight gain associated with a hypothalamic inflammation and impaired glucose homeostasis. Genetic blockade of ceramide synthesis in the ventromedial nucleus of the hypothalamus (VMH) reverses hypothalamic inflammation and partly restored glucose tolerance induced by OVX. Furthermore, glucose-stimulated insulin secretion (GSIS) is increased in OVX rats due to a raise of insulin secretion second phase, a characteristic of early stage of T2D. In contrast, GSIS from isolated islets of OVX rats is totally blunted. Inhibition of ceramide synthesis in the VMH restores GSIS from isolated OVX islets and represses the second phase of insulin secretion. Stimulation of oestrogen receptor α (ERα) by oestradiol (E2) down-regulates ceramide synthesis in hypothalamic neuronal GT1-7 cells but no in microglial SIM-A9 cells. In contrast, genetic inactivation of ERα in VMH upregulates ceramide synthesis. These results indicate that hypothalamic neuronal de novo ceramide synthesis triggers the OVX-dependent impairment of glucose homeostasis which is partly mediated by a dysregulation of GSIS.


Asunto(s)
Glucemia/fisiología , Ceramidas/biosíntesis , Hipotálamo/metabolismo , Secreción de Insulina/fisiología , Insuficiencia Ovárica Primaria/fisiopatología , Animales , Regulación hacia Abajo , Estradiol/farmacología , Femenino , Silenciador del Gen , Homeostasis , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ovariectomía , Ratas , Ratas Sprague-Dawley , Serina C-Palmitoiltransferasa/genética , Aumento de Peso
5.
Mol Metab ; 47: 101172, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33513436

RESUMEN

OBJECTIVE: Astrocytes are glial cells proposed as the main Sonic hedgehog (Shh)-responsive cells in the adult brain. Their roles in mediating Shh functions are still poorly understood. In the hypothalamus, astrocytes support neuronal circuits implicated in the regulation of energy metabolism. In this study, we investigated the impact of genetic activation of Shh signaling on hypothalamic astrocytes and characterized its effects on energy metabolism. METHODS: We analyzed the distribution of gene transcripts of the Shh pathway (Ptc, Gli1, Gli2, and Gli3) in astrocytes using single molecule fluorescence in situ hybridization combined with immunohistofluorescence of Shh peptides by Western blotting in the adult mouse hypothalamus. Based on the metabolic phenotype, we characterized Glast-CreERT2-YFP-Ptc-/- (YFP-Ptc-/-) mice and their controls over time and under a high-fat diet (HFD) to investigate the potential effects of conditional astrocytic deletion of the Shh receptor Patched (Ptc) on metabolic efficiency, insulin sensitivity, and systemic glucose metabolism. Molecular and biochemical assays were used to analyze the alteration of key pathways modulating energy metabolism, insulin sensitivity, glucose uptake, and inflammation. Primary astrocyte cultures were used to evaluate a potential role of Shh signaling in astrocytic glucose uptake. RESULTS: Shh peptides were the highest in the hypothalamic extracts of adult mice and a large population of hypothalamic astrocytes expressed Ptc and Gli1-3 mRNAs. Characterization of Shh signaling after conditional Ptc deletion in the YFP-Ptc-/- mice revealed heterogeneity in hypothalamic astrocyte populations. Interestingly, activation of Shh signaling in Glast+ astrocytes enhanced insulin responsiveness as evidenced by glucose and insulin tolerance tests. This effect was maintained over time and associated with lower blood insulin levels and also observed under a HFD. The YFP-Ptc-/- mice exhibited a lean phenotype with the absence of body weight gain and a marked reduction of white and brown adipose tissues accompanied by increased whole-body fatty acid oxidation. In contrast, food intake, locomotor activity, and body temperature were not altered. At the cellular level, Ptc deletion did not affect glucose uptake in primary astrocyte cultures. In the hypothalamus, activation of the astrocytic Shh pathway was associated with the upregulation of transcripts coding for the insulin receptor and liver kinase B1 (LKB1) after 4 weeks and the glucose transporter GLUT-4 after 32 weeks. CONCLUSIONS: Here, we define hypothalamic Shh action on astrocytes as a novel master regulator of energy metabolism. In the hypothalamus, astrocytic Shh signaling could be critically involved in preventing both aging- and obesity-related metabolic disorders.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Envejecimiento , Animales , Astrocitos/patología , Metabolismo Energético/genética , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Neuronas/metabolismo , Obesidad , Receptores Patched/deficiencia , Receptores Patched/genética , Transducción de Señal , Activación Transcripcional
6.
Cells ; 9(7)2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-32668665

RESUMEN

Obesity is a pathophysiological condition where excess free fatty acids (FFA) target and promote the dysfunctioning of insulin sensitive tissues and of pancreatic ß cells. This leads to the dysregulation of glucose homeostasis, which culminates in the onset of type 2 diabetes (T2D). FFA, which accumulate in these tissues, are metabolized as lipid derivatives such as ceramide, and the ectopic accumulation of the latter has been shown to lead to lipotoxicity. Ceramide is an active lipid that inhibits the insulin signaling pathway as well as inducing pancreatic ß cell death. In mammals, ceramide is a key lipid intermediate for sphingolipid metabolism as is sphingosine-1-phosphate (S1P). S1P levels have also been associated with the development of obesity and T2D. In this review, the current knowledge on S1P metabolism in regulating insulin signaling in pancreatic ß cell fate and in the regulation of feeding by the hypothalamus in the context of obesity and T2D is summarized. It demonstrates that S1P can display opposite effects on insulin sensitive tissues and pancreatic ß cells, which depends on its origin or its degradation pathway.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Lisofosfolípidos/metabolismo , Obesidad/metabolismo , Esfingosina/análogos & derivados , Animales , Metabolismo Energético , Humanos , Insulina/metabolismo , Mamíferos/metabolismo , Esfingosina/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-30906281

RESUMEN

Low-grade inflammation and insulin resistance are among the clinical features of obesity that are thought to promote the progressive onset of type 2 diabetes. However, the underlying mechanisms linking these disorders remain not fully understood. Recent reports pointed out hypothalamic inflammation as a major step in the onset of obesity-induced insulin resistance. In light of the increasing prevalence of obesity and T2D, two worldwide public health concerns, deciphering mechanisms implicated in hypothalamic inflammation constitutes a major challenge in the field of insulin-resistance/obesity. Several clinical and experimental studies have identified resistin as a key hormone linking insulin-resistance to obesity, notably through the activation of Toll Like Receptor (TLR) 4 signaling pathways. In this review, we present an overview of the molecular mechanisms underlying obesity-induced hypothalamic inflammation and insulin resistance with peculiar focus on the role of resistin/TLR4 signaling pathway.

8.
PLoS One ; 14(3): e0213267, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30845245

RESUMEN

Resistin promotes hypothalamic neuroinflammation and insulin resistance through Toll like receptor 4 (TLR4), this hormone is thought to be a link between obesity and insulin-resistance. Indeed, resistin plasma levels are higher in obese and insulin resistant subjects. However, the impact of maternal resistin on the predisposition of offspring to hypothalamic neuroinflammation is unknown. Here, female mice were treated with resistin during gestation/lactation periods, then hypothalamic neuroinflammation was investigated in male offspring at p28 and p90. At p28, resistin increased the expression of inflammation markers (IL6, TNFα and NFκB) and TLR4 in the hypothalamus and decreased both hypothalamic insulin and leptin receptors' expression. The hypothalamic up-regulation IL6, TNFα and TLR4 was sustained until p90 promoting most likely hypothalamic inflammation. Maternal resistin also increased IL6 and TNFα in the adipose tissue of offspring at p90 associated with a higher body weight gain. In contrast, liver and muscle were not affected. These findings reveal that the augmentation of maternal resistin during gestation and lactation promotes hypothalamic and adipose tissue inflammation of offspring as evidenced by sustained increase of inflammation markers from weaning to adulthood. Thus, maternal resistin programs offspring hypothalamic and adipose tissue inflammation predisposing then offspring to body weight gain.


Asunto(s)
Intolerancia a la Glucosa/etiología , Hipotálamo/inmunología , Inflamación/etiología , Resistencia a la Insulina , Insulinoma/etiología , Resistina/efectos adversos , Aumento de Peso/efectos de los fármacos , Animales , Animales Recién Nacidos , Peso Corporal , Femenino , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Insulinoma/metabolismo , Insulinoma/patología , Lactancia , Leptina/metabolismo , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Ratones , Embarazo , Resistina/administración & dosificación , Destete
9.
J Endocrinol ; 238(1): 77-89, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29773580

RESUMEN

Autophagy is a non-selective degradation pathway induced in energy-deprived cells and in non-starved cells by participating in cellular inflammatory responses mainly through the elimination of injured and aged mitochondria that constitute an important source of reactive oxygen species. We have previously reported that resistin/TLR4 signaling pathway induces inflammation and insulin resistance in neuronal cell. However, the impact of resistin-induced inflammation on neuronal autophagy is unknown. In the present study, we hypothesized that resistin-induced neuroinflammation could be attributed, at least partially, to the impairment of autophagy pathways in neuronal cells. Our data show that resistin decreases neuronal autophagy as evidenced by the repression of the main autophagy markers in SH-SY5Y human neuroblastoma cell line. Furthermore, the silencing of TLR4 completely abolished these effects. Resistin also inhibits AMPK phosphorylation and increases that of Akt/mTOR contrasting with activated autophagy where AMPK phosphorylation is augmented and mTOR inhibited. In vivo, resistin treatment inhibits the mRNA expression of autophagy markers in the hypothalamus of WT mice but not in Tlr4-/- mice. In addition, resistin strongly diminished LC3 (a marker of autophagy) labeling in the arcuate nucleus of WT mice, and this effect is abolished in Tlr4-/- mice. Taken together, our findings clearly reveal resistin/TLR4 as a new regulatory pathway of neuronal autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Neuronas/efectos de los fármacos , Resistina/farmacología , Receptor Toll-Like 4/fisiología , Animales , Autofagia/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Resistina/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Toll-Like 4/genética , Células Tumorales Cultivadas
10.
Endocrinology ; 147(5): 2550-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16497805

RESUMEN

The insulin-sensitive glucose transporter Glut4 is expressed in brain areas that regulate energy homeostasis and body adiposity. In contrast with peripheral tissues, however, the impact of insulin on Glut4 plasma membrane (PM) translocation in neurons is not known. In this study, we examined the role of two anorexic hormones (leptin and insulin) on Glut4 translocation in a human neuronal cell line that express endogenous insulin and leptin receptors. We show that insulin and leptin both induce Glut4 translocation to the PM of neuronal cells and activate glucose uptake. Wortmannin, a specific inhibitor of phosphatidylinositol 3-kinase, totally abolished insulin- and leptin-dependent Glut4 translocation and stimulation of glucose uptake. Thus, Glut4 translocation is a phosphatidylinositol 3-kinase-dependent mechanism in neuronal cells. Next, we investigated the impact of chronic insulin and leptin treatments on Glut4 expression and translocation. Chronic exposure of neuronal cells to insulin or leptin down-regulates Glut4 proteins and mRNA levels and abolishes the acute stimulation of glucose uptake in response to acute insulin or leptin. In addition, chronic treatment with either insulin or leptin impaired Glut4 translocation. A cross-desensitization between insulin and leptin was apparent, where exposure to insulin affects leptin-dependent Glut4 translocation and vice versa. This cross-desensitization could be attributed to the increase in suppressor of cytokine signaling-3 expression, which was demonstrated in response to each hormone. These results provide evidence to suggest that Glut4 translocation to neuronal PM is regulated by both insulin and leptin signaling pathways. These pathways might contribute to an in vivo glucoregulatory reflex involving a neuronal network and to the anorectic effect of insulin and leptin.


Asunto(s)
Membrana Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/farmacocinética , Insulina/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Androstadienos/farmacología , Transporte Biológico , Línea Celular Tumoral , Citocinas/metabolismo , Desoxiglucosa/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Modelos Biológicos , Transporte de Proteínas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factores de Tiempo , Wortmanina
11.
Biochem J ; 388(Pt 3): 929-39, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15715521

RESUMEN

Leptin and insulin are major signals to the hypothalamus to regulate energy homoeostasis and body adiposity. IR (insulin receptors) and leptin receptors (long isoform, ObRb) share a number of signalling cascades, such as JAK2/STAT-3 (Janus kinase 2/signal transduction and activator of transcription 3) and PI3K (phosphoinositide 3-kinase); the cross-talk between IR and ObRb have been described previously in non-neuronal cells. Differentiated human neuroblastoma (SH-SY5Y) cells express endogenous ObR and IR, and respond to leptin and insulin with stimulation of STAT-3 and MAPK (mitogen-activated protein kinase) phosphorylation, and PI3K activity. Insulin or leptin pre-treatment of SH-SY5Y cells increased basal STAT-3 phosphorylation, but abolished the acute effect of these hormones, and, interestingly, leptin pre-treatment abolished insulin effect and vice versa. Similar results were obtained for MAPK phosphorylation, but leptin or insulin pre-treatment did not completely abolish the acute effect of insulin or leptin. We have also showed that insulin and leptin are able to activate PI3K through IRS-1 (insulin receptor substrate 1) and IRS-2 respectively. Furthermore, leptin or insulin pre-treatment increased basal PI3K activity and IRS-1 or IRS-2 association with p85 and abolished acute insulin or leptin effect, in addition to the down-regulation of IRS-1 and IRS-2. Finally, insulin pre-treatment reduced leptin binding by approx. 60%, and leptin pre-treatment reduced the expression of insulin receptor by 40% in SH-SY5Y cells, which most likely accounts for the cross down-regulation of leptin and insulin receptors. These results provide evidence to suggest cross down-regulation of leptin and insulin receptors at both receptor and downstream signalling levels. This finding may contribute to the understanding of the complex relationship between leptin resistance and insulin resistance at the neuronal level.


Asunto(s)
Regulación hacia Abajo , Neuronas/metabolismo , Receptor de Insulina/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Inmunoprecipitación , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Leptina/metabolismo , Leptina/farmacología , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Unión Proteica , Subunidades de Proteína , Receptores de Leptina , Ovinos , Tretinoina/farmacología
13.
J Endocrinol ; 228(1): 1-12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26459640

RESUMEN

The beneficial effect of polyunsaturated omega-3 fatty acid (w-3 FA) consumption regarding cardiovascular diseases, insulin resistance and inflammation has been widely reported. Fish oil is considered as the main source of commercialized w-3 FAs, and other alternative sources have been reported such as linseed or microalgae. However, despite numerous reports, the underlying mechanisms of action of w-3 FAs on insulin resistance are still not clearly established, especially those from microalgae. Here, we report that Odontella aurita, a microalga rich in w-3 FAs eicosapentaenoic acid, prevents high fat diet-induced insulin resistance and inflammation in the liver of Wistar rats. Indeed, a high fat diet (HFD) increased plasma insulin levels associated with the impairment of insulin receptor signaling and the up-regulation of toll-like receptor 4 (TLR4) expressions. Importantly, Odontella aurita-enriched HFD (HFOA) reduces body weight and plasma insulin levels and maintains normal insulin receptor expression and responsiveness. Furthermore, HFOA decreased TLR4 expression, JNK/p38 phosphorylation and pro-inflammatory factors. In conclusion, we demonstrate for the first time, to our knowledge, that diet supplementation with whole Ondontella aurita overcomes HFD-induced insulin resistance through the inhibition of TLR4/JNK/p38 MAP kinase signaling pathways.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Dieta , Ácidos Grasos Omega-3/administración & dosificación , Resistencia a la Insulina , Hepatopatías/prevención & control , Microalgas , Adiposidad , Animales , Peso Corporal , Suplementos Dietéticos , Ácido Eicosapentaenoico/administración & dosificación , Hepatitis/prevención & control , Insulina/sangre , Lípidos/sangre , Hígado/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Fosforilación , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/antagonistas & inhibidores
14.
Diabetes ; 65(4): 913-26, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26740596

RESUMEN

Adiponectin, an insulin-sensitizing hormone, and resistin, known to promote insulin resistance, constitute a potential link between obesity and type 2 diabetes. In addition, fibroblast growth factor (FGF)21 has effects similar to those of adiponectin in regulating glucose and lipid metabolism and insulin sensitivity. However, the interplay between adiponectin, FGF21, and resistin signaling pathways during the onset of insulin resistance is unknown. Here, we investigated whether central resistin promotes insulin resistance through the impairment of adiponectin and FGF21 signaling. We show that chronic intracerebroventricular resistin infusion downregulated both hypothalamic and hepatic APPL1, a key protein in adiponectin signaling, associated with decreased Akt-APPL1 interaction and an increased Akt association with its endogenous inhibitor tribbles homolog 3. Resistin treatment also decreased plasma adiponectin levels and reduced both hypothalamic and peripheral expression of adiponectin receptors. Additionally, we report that intracerebroventricular resistin increased plasma FGF21 levels and downregulated its receptor components in the hypothalamus and peripheral tissues, promoting FGF21 resistance. Interestingly, we also show that resistin effects were abolished in TLR4 knockout mice and in neuronal cells expressing TLR4 siRNAs. Our study reveals a novel mechanism of insulin resistance onset orchestrated by a central resistin-TLR4 pathway that impairs adiponectin signaling and promotes FGF21 resistance.


Asunto(s)
Adiponectina/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Resistencia a la Insulina , Resistina/farmacología , Receptor Toll-Like 4/fisiología , Animales , Células Cultivadas , Resistencia a Medicamentos/efectos de los fármacos , Humanos , Infusiones Intraventriculares , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Wistar , Resistina/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor Toll-Like 4/genética
15.
Mol Cell Endocrinol ; 242(1-2): 59-66, 2005 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16150536

RESUMEN

Leptin resistance contributes to the pathogenesis of common obesity related metabolic diseases, including insulin resistance. However, the relationship between leptin and insulin resistance is not clearly established. Here, we show that induced hyperleptinemia by leptin infusion alters insulin signalling in rat liver. Leptin infusion clearly reduced the insulin or leptin dependent IRS-1/IRS-2 association to p85 regulatory subunit of PI 3-kinase. Leptin infusion also abolished STAT-3 phosphorylation in response to insulin or leptin and similar results were obtained for MAP-kinase phosphorylation. Hypothalamic leptin resistance was also induced by leptin infusion since leptin was unable to induce STAT-3 phosphorylation. These results provide evidence that induced hyperleptinemia can contribute to the onset of insulin resistance at least at the hepatic level.


Asunto(s)
Hipotálamo/efectos de los fármacos , Insulina/metabolismo , Leptina/metabolismo , Leptina/farmacología , Hígado/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Peso Corporal , Ingestión de Alimentos , Hipotálamo/metabolismo , Infusiones Parenterales , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intracelular , Leptina/administración & dosificación , Leptina/sangre , Hígado/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Factor de Transcripción STAT3/metabolismo , Ovinos
16.
J Endocrinol ; 226(1): 67-80, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25994005

RESUMEN

Malnutrition in the elderly is accompanied by several metabolic dysfunctions, especially alterations in energy homeostasis regulation and a loss of insulin responsiveness. Nutritional recommendations aim to enrich food with high protein and energy supplements, and protein composition and lipid quality have been widely studied. Despite the numerous studies that have examined attempts to overcome malnutrition in the elderly through such nutritional supplementation, it is still necessary to study the effects of a combination of protein, lipids, and vitamin D (VitD). This can be done in animal models of elderly malnutrition. In the present study, we investigated the effects of several diet formulae on insulin responsiveness, inflammation, and the hypothalamic expression of key genes that are involved in energy homeostasis control. To mimic elderly malnutrition in humans, elderly Wistar rats were food restricted (R, -50%) for 12 weeks and then refed for 4 weeks with one of four different isocaloric diets: a control diet; a diet where milk soluble protein (MSP) replaced casein; a blend of milk fat, rapeseed, and DHA (MRD); or a full formula (FF) diet that combined MSP and a blend of MRD (FF). All of the refeeding diets contained VitD. We concluded that: (i) food restriction led to the upregulation of insulin receptor in liver and adipose tissue accompanied by increased Tnfα in the hypothalamus; (ii) in all of the refed groups, refeeding led to similar body weight gain during the refeeding period; and (iii) refeeding with MSP and MRD diets induced higher food intake on the fourth week of refeeding, and this increase was associated with reduced hypothalamic interleukin 6 expression.


Asunto(s)
Envejecimiento/fisiología , Suplementos Dietéticos , Ingestión de Alimentos/fisiología , Hipotálamo/fisiopatología , Interleucina-6/genética , Desnutrición/dietoterapia , Leche , Anciano , Envejecimiento/genética , Envejecimiento/patología , Animales , Grasas de la Dieta/administración & dosificación , Suplementos Dietéticos/análisis , Modelos Animales de Enfermedad , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Expresión Génica , Humanos , Hipotálamo/patología , Resistencia a la Insulina , Masculino , Desnutrición/genética , Desnutrición/fisiopatología , Leche/química , Proteínas de la Leche/administración & dosificación , Ratas , Ratas Wistar , Solubilidad , Factor de Necrosis Tumoral alfa/genética , Vitamina D/administración & dosificación , Aumento de Peso
17.
Mol Cell Endocrinol ; 384(1-2): 1-11, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24394757

RESUMEN

Early in life, leptin plays a crucial role in hypothalamic neural organization. Leptin, most likely, controls neural gene expression conferring then specific phenotype regarding energy homeostasis. MicroRNAs are new regulators for several physiological functions, including the regulation of metabolism. However, the impact of leptin on hypothalamic microRNA patterns remains unknown. Here, we demonstrate that miR-200a, miR-200b and miR-429 are up-regulated in the hypothalamus of genetically obese and leptin deficient ob/ob mice. Leptin treatment down-regulates these miRNAs in ob/ob hypothalamus. The hypothalamic silencing of miR-200a increased the expression level of leptin receptor and insulin receptor substrate 2, reduced body weight gain, and restored liver insulin responsiveness. In addition, the overexpression of pre-miR-200a in a human neuroblastoma cell line impaired insulin and leptin signaling. These findings link the alteration of leptin and insulin signaling to the up-regulation of hypothalamic miR-200a which could be a new target for treatment of obesity.


Asunto(s)
Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/genética , MicroARNs/genética , Obesidad/genética , Transducción de Señal , Animales , Línea Celular Tumoral , Conducta Alimentaria , Regulación de la Expresión Génica , Humanos , Hipotálamo/fisiopatología , Proteínas Sustrato del Receptor de Insulina/agonistas , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Leptina/deficiencia , Hígado/metabolismo , Hígado/fisiopatología , Masculino , Ratones , Ratones Obesos , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Neuronas/citología , Neuronas/metabolismo , Obesidad/metabolismo , Obesidad/fisiopatología , Receptores de Leptina/agonistas , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Aumento de Peso
18.
J Clin Endocrinol Metab ; 99(8): 2821-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24758184

RESUMEN

CONTEXT: Obesity is characterized by the excessive accumulation of dysfunctional white adipose tissue (WAT), leading to a strong perturbation of metabolic regulations. However, the molecular events underlying this process are not fully understood. OBJECTIVE: MicroRNAs (miRNAs) are small noncoding RNAs acting as posttranscriptional regulators of gene expression in multiple tissues and organs. However, their expression and roles in WAT cell subtypes, which include not only adipocytes but also immune, endothelial, and mesenchymal stem cells as well as preadipocytes, have not been characterized. Design/Results: By applying differential miRNome analysis, we demonstrate that the expression of several miRNAs is dysregulated in epididymal WAT from ob/ob and high-fat diet-fed mice. Adipose tissue-specific down-regulation of miR-200a and miR-200b and the up-regulation of miR-342-3p, miR-335-5p, and miR-335-3p were observed. Importantly, a similarly altered expression of miR-200a and miR-200b was observed in obese diabetic patients. Furthermore, cell fractionation of mouse adipose tissue revealed that miRNAs are differentially expressed in adipocytes and in subpopulations from the stromal vascular fraction. Finally, integration of transcriptomic data showed that bioinformatically predicted miRNA target genes rarely showed anticorrelated expression with that of targeting miRNA, in contrast to experimentally validated target genes. CONCLUSION: Taken together, our data indicate that the dysregulated expression of miRNAs occurs in distinct cell types and is likely to affect cell-specific function(s) of obese WAT.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , MicroARNs/genética , Obesidad/genética , Tejido Adiposo Blanco/patología , Animales , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Análisis por Micromatrices , Obesidad/metabolismo , Especificidad de Órganos/genética
19.
Diabetes ; 62(1): 102-14, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22961082

RESUMEN

Resistin promotes both inflammation and insulin resistance associated with energy homeostasis impairment. However, the resistin receptor and the molecular mechanisms mediating its effects in the hypothalamus, crucial for energy homeostasis control, and key insulin-sensitive tissues are still unknown. In the current study, we report that chronic resistin infusion in the lateral cerebral ventricle of normal rats markedly affects both hypothalamic and peripheral insulin responsiveness. Central resistin treatment inhibited insulin-dependent phosphorylation of insulin receptor (IR), AKT, and extracellular signal-related kinase 1/2 associated with reduced IR expression and with upregulation of suppressor of cytokine signaling-3 and phosphotyrosine phosphatase 1B, two negative regulators of insulin signaling. Additionally, central resistin promotes the activation of the serine kinases Jun NH(2)-terminal kinase and p38 mitogen-activated protein kinase, enhances the serine phosphorylation of insulin receptor substrate-1, and increases the expression of the proinflammatory cytokine interleukin-6 in the hypothalamus and key peripheral insulin-sensitive tissues. Interestingly, we also report for the first time, to our knowledge, the direct binding of resistin to Toll-like receptor (TLR) 4 receptors in the hypothalamus, leading to the activation of the associated proinflammatory pathways. Taken together, our findings clearly identify TLR4 as the binding site for resistin in the hypothalamus and bring new insight into the molecular mechanisms involved in resistin-induced inflammation and insulin resistance in the whole animal.


Asunto(s)
Encéfalo/fisiología , Resistencia a la Insulina , Resistina/farmacología , Receptor Toll-Like 4/fisiología , Animales , Línea Celular Tumoral , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptor de Insulina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Trends Endocrinol Metab ; 21(11): 676-83, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20674387

RESUMEN

Retinoid X receptors (RXRs) have been implicated in a diversity of cellular processes ranging from cellular proliferation to lipid metabolism. These pleiotropic effects stem not only from the ability of RXRs to dimerize with diverse nuclear receptors, which exert transcriptional control on specific aspects of cell biology, but also because binding of RXR ligands to heterodimers can stimulate transcriptional activation by RXR partner receptors. This signaling network is rendered more complex by the existence of different RXR isotypes (RXRα, RXRß, RXRγ) with distinct properties that thereby modulate the transcriptional activity of RXR-containing heterodimers. This review discusses the emerging roles of RXR isotypes in the RXR signaling network and possible implications for our understanding of nuclear receptor biology and pharmacology.


Asunto(s)
Multimerización de Proteína/fisiología , Receptores X Retinoide/metabolismo , Receptores X Retinoide/fisiología , Activación Transcripcional/fisiología , Humanos , Modelos Biológicos , Modelos Moleculares , Unión Proteica/fisiología , Multimerización de Proteína/genética , Receptores X Retinoide/química , Receptores X Retinoide/genética , Especificidad por Sustrato , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA